Browsing by Subject "immunotherapy"

Sort by: Order: Results:

Now showing items 1-20 of 38
  • Clubb, James H. A.; Kudling, Tatiana V.; Heinioe, Camilla; Basnet, Saru; Pakola, Santeri; Cervera Carrascon, Victor; Santos, Joao Manuel; Quixabeira, Dafne C. A.; Havunen, Riikka; Sorsa, Suvi; Zheng, Vincent; Salo, Tuula; Bäck, Leif; Aro, Katri; Tulokas, Sanni; Loimu, Venla; Hemminki, Akseli (2022)
    Immune checkpoint inhibitors (ICI) have provided significant improvement in clinical outcomes for some patients with solid tumors. However, for patients with head and neck cancer, the response rate to ICI monotherapy remains low, leading to the exploration of combinatorial treatment strategies. In this preclinical study, we use an oncolytic adenovirus (Ad5/3) encoding hTNF alpha and hIL-2 and non-replicate adenoviruses (Ad5) encoding mTNF alpha and mIL-2 with ICI to achieve superior tumor growth control and improved survival outcomes. The in vitro effect of Ad5/3-E2F-D24-hTNFa-IRES-hIL-2 was characterized through analyses of virus replication, transgene expression and lytic activity using head and neck cancer patient derived cell lines. Mouse models of ICI naive and refractory oral cavity squamous cell carcinoma were established to evaluate the local and systemic anti-tumor immune response upon ICI treatment with or without the non-replicative adenovirus encoding mTNF alpha and mIL-2. We delineated the mechanism of action by measuring the metabolic activity and effector function of CD3(+) tumor infiltrating lymphocytes (TIL) and transcriptomic profile of the CD45(+) tumor immune compartment. Ad5/3-E2F-D24-hTNFa-IRES-hIL-2 demonstrated robust replicative capability in vitro across all head and neck cell lines screened through potent lytic activity, E1a and transgene expression. In vivo, in both ICI naive and refractory models, we observed improvement to tumor growth control and long-term survival when combining anti-PD-1 or anti-PD-L1 with the non-replicative adenovirus encoding mTNF alpha and mIL-2 compared to monotherapies. This observation was verified by striking CD3(+) TIL derived mGranzyme b and interferon gamma production complemented by increased T cell bioenergetics. Notably, interrogation of the tumor immune transcriptome revealed the upregulation of a gene signature distinctive of tertiary lymphoid structure formation upon treatment of murine anti-PD-L1 refractory tumors with non-replicative adenovirus encoding mTNF alpha and mIL-2. In addition, we detected an increase in anti-tumor antibody production and expansion of the memory T cell compartment in the secondary lymphoid organs. In summary, a non-replicative adenovirus encoding mTNF alpha and mIL-2 potentiates ICI therapy, demonstrated by improved tumor growth control and survival in head and neck tumor-bearing mice. Moreover, the data reveals a potential approach for inducing tertiary lymphoid structure formation. Altogether our results support the clinical potential of combining this adenovirotherapy with anti-PD-1 or anti-PD-L1.
  • Sousa-Pinto, Bernardo; Azevedo, Luis Filipe; Sa-Sousa, Ana; Vieira, Rafael Jose; Amaral, Rita; Klimek, Ludger; Czarlewski, Wienczyslawa; Anto, Josep M.; Bedbrook, Anna; Kvedariene, Violeta; Ventura, Maria Teresa; Ansotegui, Ignacio J.; Bergmann, Karl-Christian; Brussino, Luisa; Canonica, G. Walter; Cardona, Victoria; Carreiro-Martins, Pedro; Casale, Thomas; Cecchi, Lorenzo; Chivato, Tomas; Chu, Derek K.; Cingi, Cemal; Costa, Elisio M.; Cruz, Alvaro A.; De Feo, Giulia; Devillier, Philippe; Fokkens, Wytske J.; Gaga, Mina; Gemicioglu, Bilun; Haahtela, Tari; Ivancevich, Juan Carlos; Ispayeva, Zhanat; Jutel, Marek; Kuna, Piotr; Kaidashev, Igor; Kraxner, Helga; Larenas-Linnemann, Desiree E.; Laune, Daniel; Lipworth, Brian; Louis, Renaud; Makris, Michael; Monti, Riccardo; Morais-Almeida, Mario; Moesges, Ralph; Mullol, Joaquim; Odemyr, Mikaela; Okamoto, Yoshitaka; Papadopoulos, Nikolaos G.; Patella, Vincenzo; Nhan Pham-Thi; Regateiro, Frederico S.; Reitsma, Sietze; Rouadi, Philip W.; Samolinski, Boleslaw; Sova, Milan; Todo-Bom, Ana; Taborda-Barata, Luis; Tomazic, Peter Valentin; Toppila-Salmi, Sanna; Sastre, Joaquin; Tsiligianni, Ioanna; Valiulis, Arunas; Wallace, Dana; Waserman, Susan; Yorgancioglu, Arzu; Zidarn, Mihaela; Zuberbier, Torsten; Fonseca, Joao Almeida; Bousquet, Jean; Pfaar, Oliver (2022)
    Background Evidence regarding the effectiveness of allergen immunotherapy (AIT) on allergic rhinitis has been provided mostly by randomised controlled trials, with little data from real-life studies. Objective To compare the reported control of allergic rhinitis symptoms in three groups of users of the MASK-air(R) app: those receiving sublingual AIT (SLIT), those receiving subcutaneous AIT (SCIT), and those receiving no AIT. Methods We assessed the MASK-air(R) data of European users with self-reported grass pollen allergy, comparing the data reported by patients receiving SLIT, SCIT and no AIT. Outcome variables included the daily impact of allergy symptoms globally and on work (measured by visual analogue scales-VASs), and a combined symptom-medication score (CSMS). We applied Bayesian mixed-effects models, with clustering by patient, country and pollen season. Results We analysed a total of 42,756 days from 1,093 grass allergy patients, including 18,479 days of users under AIT. Compared to no AIT, SCIT was associated with similar VAS levels and CSMS. Compared to no AIT, SLIT-tablet was associated with lower values of VAS global allergy symptoms (average difference = 7.5 units out of 100; 95% credible interval [95%CrI] = -12.1;-2.8), lower VAS Work (average difference = 5.0; 95%CrI = -8.5;-1.5), and a lower CSMS (average difference = 3.7; 95%CrI = -9.3;2.2). When compared to SCIT, SLIT-tablet was associated with lower VAS global allergy symptoms (average difference = 10.2; 95%CrI = -17.2;-2.8), lower VAS Work (average difference = 7.8; 95%CrI = -15.1;0.2), and a lower CSMS (average difference = 9.3; 95%CrI = -18.5;0.2). Conclusion In patients with grass pollen allergy, SLIT-tablet, when compared to no AIT and to SCIT, is associated with lower reported symptom severity. Future longitudinal studies following internationally-harmonised standards for performing and reporting real-world data in AIT are needed to better understand its 'real-world' effectiveness.
  • Bousquet, Jean; Pfaar, Oliver; Agache, Ioana; Bedbrook, Anna; Akdis, Cezmi A.; Canonica, G. Walter; Chivato, Tomas; Al-Ahmad, Mona; Abdul Latiff, Amir H.; Ansotegui, Ignacio J.; Bachert, Claus; Baharuddin, Abdullah; Bergmann, Karl-Christian; Bindslev-Jensen, Carsten; Bjermer, Leif; Bonini, Matteo; Bosnic-Anticevich, Sinthia; Bosse, Isabelle; Brough, Helen A.; Brussino, Luisa; Calderon, Moises A.; Caraballo, Luis; Cardona, Victoria; Carreiro-Martins, Pedro; Casale, Tomas; Cecchi, Lorenzo; Cepeda Sarabia, Alfonso M.; Chkhartishvili, Ekaterine; Chu, Derek K.; Cirule, Ieva; Cruz, Alvaro A.; Czarlewski, Wienczyslawa; del Giacco, Stefano; Demoly, Pascal; Devillier, Philippe; Dokic, Dejan; Durham, Stephen L.; Ebisawa, Motohiro; El-Gamalt, Yehia; Emuzyte, Regina; Gamkrelidze, Amiran; Fauquert, Jean Luc; Fiocchi, Alessandro; Fokkens, Wytske J.; Fonseca, Joao A.; Fontaine, Jean-Francois; Gawlik, Radoslaw; Gelincik, Asli; Gemicioglu, Bilun; Gereda, Jose E.; Gerth van Wijk, Roy; Gomez, R. Maximiliano; Gotua, Maia; Grisle, Ineta; Guzman, Maria-Antonieta; Haahtela, Tari; Halken, Susanne; Heffler, Enrico; Hoffmann-Sommergruber, Karin; Hossny, Elham; Hrubisko, Martin; Irani, Carla; Ivancevich, Juan Carlos; Ispayeva, Zhanat; Julge, Kaja; Kaidashev, Igor; Kalayci, Omer; Khaitov, Musa; Klimek, Ludger; Knol, Edward; Kowalski, Marek L.; Kraxner, Helga; Kull, Inger; Kuna, Piotr; Kvedariene, Violeta; Kritikos, Vicky; Lauerma, Antti; Lau, Susanne; Laune, Daniel; Levin, Michael; Larenas-Linnemann, Desiree E.; Lodrup Carlsen, Karin C.; Lombardi, Carlo; Lourenco, Olga M.; Mahboub, Bassam; Malling, Hans-Jorgen; Manning, Patrick; Marshall, Gailen D.; Melen, Erik; Meltzer, Eli O.; Miculinic, Neven; Milenkovic, Branislava; Moin, Mostafa; Montefort, Stephen; Morais-Almeida, Mario; Mortz, Charlotte G.; Mosges, Ralph; Mullol, Joaquim; Namazova Baranova, Leyla; Neffen, Hugo; Nekam, Kristof; Niedoszytko, Marek; Odemyr, Mikaela; O'Hehir, Robyn E.; Ollert, Markus; O'Mahony, Liam; Ohta, Ken; Okamoto, Yoshitaka; Okubo, Kimi; Pajno, Giovanni B.; Palomares, Oscar; Palkonen, Susanna; Panzner, Petr; Papadopoulos, Nikolaos; Park, Hae-Sim; Passalacqua, Giovanni; Patella, Vincenzo; Pawankar, Ruby; Pham-Thi, Nhan; Plavec, Davor; Popov, Todor A.; Recto, Marysia; Regateiro, Frederico S.; Riggioni, Carmen; Roberts, Graham; Rodriguez-Gonzales, Monica; Rosario, Nelson; Rottem, Menachem; Rouadi, Philip W.; Ryan, Dermot; Samolinski, Boleslaw; Sanchez-Borgest, Mario; Serpa, Faradiba S.; Sastre, Joaquin; Scadding, Glenis K.; Shamji, Mohamed H.; Schmid-Grendelmeier, Peter; Schunemann, Holger J.; Sheikh, Aziz; Scichilone, Nicola; Sisul, Juan Carlos; Sofiev, Mikhail; Sole, Dirceu; Sooronbaev, Talant; Soto-Martinez, Manuel; Soto-Quiros, Manuel; Sova, Milan; Schwarze, Jurgen; Skypala, Isabel; Suppli-Ulrik, Charlotte; Taborda-Barata, Luis; Todo-Bom, Ana; Torres, Maria J.; Valentin-Rostan, Marylin; Tomazic, Peter-Valentin; Valero, Antonio; Toppila-Salmi, Sanna; Tsiligianni, Ioanna; Untersmayr, Eva; Urrutia-Pereira, Marilyn; Valiulis, Arunas; Valovirta, Erkka; Vandenplas, Olivier; Ventura, Maria Teresa; Vichyanond, Pakit; Wagenmann, Martin; Wallace, Dana; Walusiak-Skorupa, Jolanta; Wang, De Yun; Waserman, Susan; Wong, Gary W. K.; Yorgancioglu, Arzu; Yusuf, Osman M.; Zernotti, Mario; Zhang, Luo; Zidarn, Mihaela; Zuberbier, Torsten; Jutel, Marek (2021)
  • Fusciello, Manlio; Fontana, Flavia; Tähtinen, Siri; Capasso, Cristian; Feola, Sara; da Silva Lopes Martins, Beatriz; Chiaro, Jacopo; Peltonen, Karita; Ylösmäki, Leena; Ylösmäki, Erkko; Hamdan Hissaoui, Firas; Kari, Otto K.; Ndika, Joseph; Alenius, Harri; Urtti, Arto; Hirvonen, Jouni T.; Santos, Hélder A.; Cerullo, Vincenzo (2019)
    Virus-based cancer vaccines are nowadays considered an interesting approach in the field of cancer immunotherapy, despite the observation that the majority of the immune responses they elicit are against the virus and not against the tumor. In contrast, targeting tumor associated antigens is effective, however the identification of these antigens remains challenging. Here, we describe ExtraCRAd, a multi-vaccination strategy focused on an oncolytic virus artificially wrapped with tumor cancer membranes carrying tumor antigens. We demonstrate that ExtraCRAd displays increased infectivity and oncolytic effect in vitro and in vivo. We show that this nanoparticle platform controls the growth of aggressive melanoma and lung tumors in vivo both in preventive and therapeutic setting, creating a highly specific anti-cancer immune response. In conclusion, ExtraCRAd might serve as the next generation of personalized cancer vaccines with enhanced features over standard vaccination regimens, representing an alternative way to target cancer.
  • Fontana, Flavia; Fusciello, Manlio; Groeneveldt, Christianne; Capasso, Cristian; Chiaro, Jacopo; Feola, Sara; Liu, Zehua; Mäkilä, Ermei; Salonen, Jarno; Hirvonen, Jouni; Cerullo, Vincenzo; Santos, Hélder A. (2019)
    Recent approaches in the treatment of cancer focus on involving the immune system to control the tumor growth. The administration of immunotherapies, like checkpoint inhibitors, has shown impressive results in the long term survival of patients. Cancer vaccines are being investigated as further tools to prime tumor-specific immunity. Biomaterials show potential as adjuvants in the formulation of vaccines, and biomimetic elements derived from the membrane of tumor cells may widen the range of antigens contained in the vaccine. Here, we show how mice presenting an aggressive melanoma tumor model treated twice with the complete nanovaccine formulation showed control on the tumor progression, while in a less aggressive model, the animals showed remission and control on the tumor progression, with a modification in the immunological profile of the tumor microenvironment. We also prove that co-administration of the nanovaccine together with a checkpoint inhibitor increases the efficacy of the treatment (87.5% of the animals responding, with 2 remissions) compared to the checkpoint inhibitor alone in the B16.OVA model. Our platform thereby shows potential applications as a cancer nanovaccine in combination with the standard clinical care treatment for melanoma cancers.
  • Almangush, Alhadi; Leivo, Ilmo; Mäkitie, Antti A. (2021)
    Oral squamous cell carcinoma (OSCC) forms a major health problem in many countries. For several decades the management of OSCC consisted of surgery with or without radiotherapy or chemoradiotherapy. Aiming to increase survival rate, recent research has underlined the significance of harnessing the immune response in treatment of many cancers. The promising finding of checkpoint inhibitors as a weapon for targeting metastatic melanoma was a key event in the development of immunotherapy. Furthermore, clinical trials have recently proven inhibitor of PD-1 for treatment of recurrent/metastatic head and neck cancer. However, some challenges (including patient selection) are presented in the era of immunotherapy. In this mini-review we discuss the emergence of immunotherapy for OSCC and the recently introduced biomarkers of this therapeutic strategy. Immune biomarkers and their prognostic perspectives for selecting patients who may benefit from immunotherapy are addressed. In addition, possible use of such biomarkers to assess the response to this new treatment modality of OSCC will also be discussed.
  • Kozlova, Anastasia (Helsingin yliopisto, 2023)
    Chimeric antigen receptor (CAR) T cells are genetically modified usually autologous T cells expressing de novo designed CAR that binds a specific antigen on the surface of the cancer cells, inducing T cell receptor-independent activation and cytotoxic response against the targeted cancer cells. While CAR T cells have been shown to offer effective treatment in acute lymphoblastic leukemia, diffuse large B-cell lymphoma, and multiple myeloma, several resistance mechanisms can lead to CAR T cell exhaustion characterized by impaired functions and the expression of inhibitory receptors. The Finnish Red Cross Blood Service has developed novel CARs, differing in structure from the ones currently published. Since the evasion of CAR T cell exhaustion is considered one of the key objectives in the development of CAR T cell therapy, this Master’s thesis project aimed to create a working method to determine the exhaustion of CAR T cells in vitro after long-term repeated stimulation. In order to induce and measure exhaustion, CAR T cells were produced and activated ex vivo in the presence of IL-2 or IL-7/IL-15 cytokines, cultured long-term and repeatedly stimulated by exposure to target cells. CAR T cell cytotoxicity and expansion were determined and the expression of various inhibitory receptors was analyzed. The method enabled the comparison of the designed CAR T cell candidates and the positive control CD19-CD28ζ CAR T cells in long-term cytotoxic potency. In addition, it helped to reveal the surprising difference between IL-2 and IL-7/IL-15 cytokines and their impact on CAR T cell exhaustion. Although CAR T cells produced with IL-2 had poorer expansion during CAR T cell production than CAR T cells produced with IL-7/IL-15, they showed lower expression of exhaustion-related markers supported by better survival, proliferation and cytotoxic activity during long-term repeated stimulation assay.
  • Harjunpää, Heidi; Llort Asens, Marc; Guenther, Carla; Fagerholm, Susanna C. (2019)
    The immune system and cancer have a complex relationship with the immune system playing a dual role in tumor development. The effector cells of the immune system can recognize and kill malignant cells while immune system-mediated inflammation can also promote tumor growth and regulatory cells suppress the anti-tumor responses. In the center of all anti-tumor responses is the ability of the immune cells to migrate to the tumor site and to interact with each other and with the malignant cells. Cell adhesion molecules including receptors of the immunoglobulin superfamily and integrins are of crucial importance in mediating these processes. Particularly integrins play a vital role in regulating all aspects of immune cell function including immune cell trafficking into tissues, effector cell activation and proliferation and the formation of the immunological synapse between immune cells or between immune cell and the target cell both during homeostasis and during inflammation and cancer. In this review we discuss the molecular mechanisms regulating integrin function and the role of integrins and other cell adhesion molecules in immune responses and in the tumor microenvironment. We also describe how malignant cells can utilize cell adhesion molecules to promote tumor growth and metastases and how these molecules could be targeted in cancer immunotherapy.
  • Mäki, Toni (Helsingin yliopisto, 2020)
    The human immune system can provide a powerful tool in developing therapies against various cancers. Even though the idea of an immune system actively searching for and disposing of potential mutated tumor cells is over a century old, only recent developments in various fields such as mass spectrometry, immuno-checkpoint blockade strategies and in silico modelling have enabled the realization of the full potential of recruiting immune system to fight cancer and the possibilities of personalized therapies. These therapeutic methods, including but not limited to oncolytic virus therapies, T-cell therapies and cancer vaccines, are based on the body’s ability to recognize mutated antigen peptides presented on the cell surface by MCH-receptors (also known as HLA-receptors in humans) and the disposal of the malignant cells by cytotoxic T-cells. Thus, the capability to map the individual HLA-presented peptidome and differentiate the immunogenic peptides is a foundation for this plethora of therapies and is in focus of ongoing research. This master thesis is a part of a project aiming to set up immunoaffinity-purification/MS based method in order to analyse the ligandome and determine T-cell recognized cancer associated antigens from tumor cells. Objectives of the work: 1. Characterizing tumor cell lines. 2. Immunological assay set up. 3. Collecting cell culture material for the ligandome affinity purification. 4. In silico prediction if the immunogenicity of selected peptides and assessing their source proteins. Methods used: 1. Cell culture. 2. FACS-analysis. 3. MTS-viability assay. 4. Immunological assays (ELISA, ELISPOT). 5. Immunological bioinformatics analysis tools (IEDB) and database search (UniPROT). Results: 1. Flow cytometric analysis provided essential information of the cell line HLA-1 expression. Additional information of PD-L1 expression can be used to evaluate cell line’s immune-evasion abilities. Preliminary MTS assay is used to determine linear range and optimal time frame for the PBMC/cancer cell co-culture killing assay. 2. Interferon γ cytokine secretion was determined by ELISPOT to assess PBMC response against known antigens in a preliminary experiment to approximate usable range for the following antigen specific PBMC assays. ELISA is used to confirm the presence of HLA-I receptors in the ligandome affinity purification eluates and to estimate the efficacy of purification. 3. Feasibility of in silico methods in the prediction of immunogenic peptides was explored. The experiments provided information that can be applied to the further development of the immune ligandome discovery project. In silico methods were successfully used to characterize previously identified HLA-restricted peptides and one previously identified immunogenic T-cell epitope. Even if the data acquired in silico can be considered only nominally verified at this stage, the results are encouraging.
  • Havunen, Riikka; Kalliokoski, Riikka; Siurala, Mikko; Sorsa, Suvi; Santos, Joao M.; Cervera-Carrascon, Victor; Anttila, Marjukka; Hemminki, Akseli (2021)
    Oncolytic viruses provide a biologically multi-faceted treatment option for patients who cannot be cured with currently available treatment options. We constructed an oncolytic adenovirus, TILT-123, to support T-cell therapies and immune checkpoint inhibitors in solid tumors. Adenoviruses are immunogenic by nature, are easy to produce in large quantities, and can carry relatively large transgenes. They are the most commonly used gene therapy vectors and are well tolerated in patients. TILT-123 expresses two potent cytokines, tumor necrosis factor alpha and interleukin-2, to stimulate especially the T-cell compartment in the tumor microenvironment. Before entering clinical studies, the safety and biodistribution of TILT-123 was studied in Syrian hamsters and in mice. The results show that TILT-123 is safe in animals as monotherapy and in combination with an immune checkpoint inhibitor anti-PD-1. The virus treatment induces acute changes in circulating immune cell compartments, but the levels return to normal by the middle of the treatment period. The virus is rapidly cleared from healthy tissues, and it does not cause damage to vital organs. The results support the initiation of a phase 1 dose-escalation trial, where melanoma patients receiving a tumor-infiltrating lymphocyte therapy are treated with TILT-123 (NCT04217473).
  • Juntunen, Maiju (Helsingin yliopisto, 2020)
    Cancer immunotherapy refers to therapy strategies that utilise the mechanisms of the immune system to treat cancer patients. The benefits of the approach include the possibility for specific targeting and utilisation of the host immune system. The treatment methods include cancer vaccines, oncolytic viruses (OVs), cell-based immunotherapies and antibodies. The interplay between the cancer and the immune system has been observed crucial for the progress of the cancer and the success of immunotherapies. An immune inflamed tumour microenvironment has been observed beneficial for the success of several therapy methods. Many immunotherapy methods rely on detecting tumour specific antigens that are used to guide the therapy agent to the target site. This strategy poses challenges when considering tumour immune evasion mechanisms, which can cause downregulation of target antigens, and heterogeneity of tumour cells and patients. OVs have the advantage of not requiring predetermined target structures to exert their effect to the tumour cells. They cause direct tumour cell lysis and induce immune responses, and may be modified to express additional genes, including immunostimulatory agents. However, virus-related immunosuppressive mechanisms and a rapid viral clearance may limit their effects. A Western Reserve (WR) Vaccinia virus (VACV) is a highly oncolytic virus strain but the virus has been observed to suppress the function of the cyclic guanosine monophosphate adenosine monophosphate synthase – stimulator of interferon genes (cGAS STING) innate immune pathway which has been shown to have a significant role in anti-tumour immune responses. The aim of this study was to create a WR VACV encoding a dominantly active (D A) STING and to determine whether the virus is capable of activating the cGAS STING pathway. The effects were compared to a corresponding virus vvdd tdTomato that does not have the STING encoding gene. The pathogenicity of viruses was controlled by a double deletion of the thymidine kinase and vaccinia growth factor genes which restricts the virus replication to tumour cells. Transgene fragments were cloned from template plasmids by polymerase chain reactions (PCRs) and joined together in a Gibson Assembly (GA) reaction to form a STING-P2A-eGFP gene insert. The insert was attached to a shuttle vector pSC65-tdTomato by restriction enzyme digestion, ligation and transformation in Escherichia coli. The correct transgene plasmid construct was verified by Sanger sequencing and PCRs. The transgene was inserted to a modified WR VACV vvdd-tdTomato-hDAI by a homologous recombination. The newly created VVdd STING-P2A-eGFP virus was purified by plaque purification. The STING protein expression was studied by an immunocytochemistry (ICC) assay. The immune signalling pathway activation was examined by testing nuclear factor kappa-light chain-enhancer of activated B cells (NF-κB) activation in RAW-Blue cells and dendritic cell activation and maturation in JAWS II cells. The cell viability after iinfection was studied with four cell lines; A549, B16-F10, HEK293 and MB49. The D-A STING expressing virus was produced successfully. The ICC experiment verified the capability of the VVdd STING-P2A eGFP to produce the STING protein in the infected cells. The preliminary findings indicate that the VVdd STING-P2A-eGFP virus activates the NF-κB signalling in the RAW-Blue cells and that the activation is dependent on the STING expression. The activation level is relative to the infection concentration at MOI range 0,001 to 0,1. The findings suggest that the VVdd-STING-eGFP virus can induce innate immune signalling via the STING pathway. The reference virus did not activate the signalling. The in vitro experiments also indicated that the STING virus may induce DC activation and maturation. We observed a trend of CD86 and CD40 expression upregulation on the JAWS II DCs. The effects to the cell viability were inconclusive. More studies should be conducted to verify the results. The effects of the virus should be studied in more advanced cancer models that take into account the complexity of the immune system. These preliminary results indicate the that the VVdd-STING-P2A-eGFP virus could stimulate the immune signalling through the STING pathway.
  • Jansen, Y. J. L.; Rozeman, E. A.; Mason, R.; Goldinger, S. M.; Foppen, M. H. Geukes; Hoejberg, L.; Schmidt, H.; van Thienen, J.; Haanen, J. B. A. G.; Tiainen, L.; Svane, I. M.; Mäkelä, S.; Seremet, T.; Arance, A.; Dummer, R.; Bastholt, L.; Nyakas, M.; Straume, O.; Menzies, A. M.; Long, G.; Atkinson, Jeffrey; Blank, C. U.; Neyns, B. (2019)
    Background Programmed cell death protein 1 (PD-1) blocking monoclonal antibodies improve the overall survival of patients with advanced melanoma but the optimal duration of treatment has not been established. Patients and Methods This academic real-world cohort study investigated the outcome of 185 advanced melanoma patients who electively discontinued anti-PD-1 therapy with pembrolizumab (N=167) or nivolumab (N=18) in the absence of disease progression (PD) or treatment limiting toxicity (TLT) at 14 medical centres across Europe and Australia. Results Median time on treatment was 12months (range 0.7-43). The best objective tumour response at the time of treatment discontinuation was complete response (CR) in 117 (63%) patients, partial response (PR) in 44 (24%) patients and stable disease (SD) in 16 (9%) patients; 8 (4%) patients had no evaluable disease (NE). After a median follow-up of 18months (range 0.7-48) after treatment discontinuation, 78% of patients remained free of progression. Median time to progression was 12months (range 2-23). PD was less frequent in patients with CR (14%) compared with patients with PR (32%) and SD (50%). Six out of 19 (32%) patients who were retreated with an anti-PD-1 at the time of PD obtained a new antitumour response. Conclusions In this real-world cohort of advanced melanoma patients discontinuing anti-PD-1 therapy in the absence of TLT or PD, the duration of anti-PD-1 therapy was shorter when compared with clinical trials. In patients obtaining a CR, and being treated for >6months, the risk of relapse after treatment discontinuation was low. Patients achieving a PR or SD as best tumour response were at higher risk for progression after discontinuing therapy, and defining optimal treatment duration in such patients deserves further study. Retreatment with an anti-PD-1 at the time of progression may lead to renewed antitumour activity in some patients. Clinical trial registration NCT02673970 (https://clinicaltrials.gov/ct2/show/NCT02673970?cond=melanoma&cntry=BE&city=Jette&rank=3)
  • Mahlamäki, Kasper (Helsingin yliopisto, 2019)
    Syöpätautien yleisyys lisääntyy kaikkialla maailmassa väestön vanhetessa ja Suomessakin todetaan vuosittain noin 35 000 uutta syöpää, joihin kuolee noin 13 000 henkeä. Syövän lääkehoidon painopiste on siirtymässä laajalti elimistön soluja tuhoavista sädehoidosta ja solunsalpaajista kohti potilaan omaa immuunipuolustusta syöpää vastaan ohjaavia immuno-onkologisia lääkkeitä. Immuno-onkologisilla lääkkeillä, kuten tarkastuspiste-estäjiin kuuluvilla PD-1-vasta-aineilla on saavutettu jopa pysyviä remissioita useassa vaikeahoitoisessa syövässä. Valitettavasti vain pieni osa immunologisilla syöpälääkkeillä hoidetuista potilaista saa niiden täyden hyödyn ja hoitovasteen laajentaminen koko potilasjoukkoon on osoittautunut ongelmalliseksi. Tutkielmani kirjallisuuskatsauksessa käsitellään rintasyöpää yleisesti, sekä syvennytään rintasyövän prekliinisiin tutkimusmalleihin ja rintasyöpäpotilaan immuno-onkologiavasteen ennustetekijöihin. Kokeellisen laboratoriotutkimusosuuden tavoitteina oli korreloida rintasyöpänäytteiden leukosyytti-infiltraation taso kasvaimen Myc- ja PD-L1-ekspressioon, mitata solulinjojen ja tutkimusryhmämme kehittämien PDEC-eksplanttien immuuniaktivaatiota, sekä arvioida näiden tutkimusten tulosten avulla rintasyöpäpotilaan kasvainnäytteestä kasvattamiemme PDEC-kudosviljelmien potentiaalia rintasyövän immuno-onkologisessa tutkimuksessa. Löysin tutkimalla primäärikasvainten ja PDEC-eksplanttien ominaisuuksia immunohistokemialla sekä qRT-PCR:llä korrelaation primäärikasvaimen leukosyyttimäärän sekä PDEC-viljelmien qRT-PCR:llä mitatun immuuniaktiivisuuden välille: kasvaimen korkea leukosyyttipitoisuus vaikuttaa olevan yhteydessä PDEC-viljelmien suurempaan immuuniaktiivisuuteen. Mielenkiintoista oli havaita, että vaikka immuunisolujen aktivoimiseen käytetyn Immunocultin tulisi aktivoida kaikkia T-soluja, niin edes runsas kasvaimen strooman leukosyytti-infiltraatio ei aiheuttanut qRT-PCR:llä tehdyissä immuuniaktivaatiomittauksissa menetelmän käyttöä häiritseviä vääriä positiivisia tuloksia. Tulosteni perusteella PDEC-eksplantit kykenevät kuvaamaan rintasyöpäpotilaiden kasvainkohtaisia immuuniaktivaatioeroja, mikä korostaa niiden soveltuvuutta immuno-onkologiseen tutkimukseen.
  • Bouhlal, Jonas Otto Vilhelm (Helsingin yliopisto, 2022)
    Despite of great advancements in the field of cancer therapy in the past decades, the 5-year survival of acute myeloid leukaemia (AML) patients remains low with high mortality especially in elderly patients, in whom the disease is most often observed. Poor prognosis often results from complex heterogenous molecular abnormalities defining the progress of the disease, while making it more difficult to treat due to intensive treatments only being feasible for younger patients. Our increased understanding of cancer immunology and the potential of immunotherapy has, however, led to promising therapeutic innovations, which give hope for discovering long-lasting and effective treatment options. Natural killer (NK) cell-based immunotherapies are amongst the emerging novel therapeutic approaches that aim to target malignant cells with less toxicity and improved applicability. Using high-throughput drug sensitivity and resistance testing combined with single cell RNA (scRNA) sequencing, this study focused on finding drug compounds that could synergise with NK cells to improve their effectiveness in killing leukemic cells. In this study, many drugs showed promising results in being able to potentiate NK cell cytotoxicity, with daporinad and pevonedistat showing the most notable differences when compared to controls. The potentiating effect of Janus kinase (JAK) inhibitors also suggested a method of increasing NK cell activity against leukemic cells through downregulation of major histocompatibility complex (MHC) class I molecules. In conclusion, findings shed light on the synergetic potential of drugs and NK cells, giving hope for clinically relevant findings following further validation and testing.
  • Sieviläinen, Meri; Saavalainen, Jordan; Adnan-Awad, Shady; Salo, Tuula; Al-Samadi, Ahmed (2022)
    BackgroundImmune checkpoint inhibitors (ICIs), primarily anti-PD-1, are currently used to treat patients with recurrent or metastatic head and neck squamous cell carcinoma (HNSCC). However, only a minority of patients benefit from these costly therapies. Therefore, there is an unmet need to better understand the effect of ICIs on immune effector cells. This study aimed to investigate the effect of a PD-1 antibody and an IDO1 inhibitor on different lymphocyte populations (NK, CD4(+), and CD8(+) T cells) in term of migration, cytotoxicity, and cytokine release in the presence of HNSCC cells. MethodsUsing a microfluidic chip, we injected HSC-3 cells (an oral tongue squamous cell carcinoma cell line) embedded in a human tumor-derived matrix "myogel/fibrin" together with NK, CD4(+), and CD8(+) T cells in separate channels. The two channels were connected with microchannels. The PD-1 antibody nivolumab and IDO1 inhibitor epacadostat were added to the microfluidic chips. Lymphocyte migration and cytotoxicity were examined under fluorescent microscopy and cytokine release was measured using a FirePlex Human Discovery Cytokines Immunoassay. ResultsEpacadostat significantly increased the migration and infiltration of NK and CD4(+) T cells, but not CD8(+) T cells, towards the cancer cells. Nivolumab did not exhibit a similar effect. While CD8(+) T cells alone showed near to no migration, adding CD4(+) T cells enhanced migration towards the cancer cells. There was a mild nonsignificant increase in apoptosis of HSC-3 cells after adding epacadostat to lymphocytes. In contrast, HSC-3 proliferation was not affected by lymphocytes regardless of ICIs. Nivolumab significantly increased release of MIP1-alpha, IL-6, and IL-8 from NK, CD4(+), and CD8(+) T cells, respectively. ConclusionsThis study revealed that each subpopulation of lymphocytes respond differently to ICIs. We also revealed the subpopulation of lymphocytes responsible for the increases in specific serum cytokines after ICI treatment.
  • Hemminki, Otto; Parviainen, Suvi; Juhila, Juuso; Turkki, Riku; Linder, Nina; Lundin, Johan; Kankainen, Matti; Ristimaki, Ari; Koski, Anniina; Liikanen, Ilkka; Oksanen, Minna; Nettelbeck, Dirk M.; Kairemo, Kalevi; Partanen, Kaarina; Joensuu, Timo; Kanerva, Anna; Hemminki, Akseli (2015)
    Oncolytic viruses that selectively replicate in tumor cells can be used for treatment of cancer. Accumulating data suggests that virus induced oncolysis can enhance anti-tumor immunity and break immune tolerance. To capitalize on the immunogenic nature of oncolysis, we generated a quadruple modified oncolytic adenovirus expressing granulocyte-macrophage colony-stimulating factor (GMCSF). Ad5/3-E2F-Delta 24-GMCSF (CGTG-602) was engineered to contain a tumor specific E2F1 promoter driving an E1 gene deleted at the retinoblastoma protein binding site ("Delta 24"). The fiber features a knob from serotype 3 for enhanced gene delivery to tumor cells. The virus was tested preclinically in vitro and in vivo and then 13 patients with solid tumors refractory to standard therapies were treated. Treatments were well tolerated and frequent tumor-and adenovirus-specific T-cell immune responses were seen. Overall, with regard to tumor marker or radiological responses, signs of antitumor efficacy were seen in 9/12 evaluable patients (75%). The radiological disease control rate with positron emission tomography was 83% while the response rate (including minor responses) was 50%. Tumor biopsies indicated accumulation of immunological cells, especially T-cells, to tumors after treatment. RNA expression analyses of tumors indicated immunological activation and metabolic changes secondary to virus replication.
  • Kudling, Tatiana V.; Clubb, James H. A.; Quixabeira, Dafne C. A.; Santos, Joao M.; Havunen, Riikka; Kononov, Alexander; Heiniö, Camilla; Cervera-Carrascon, Victor; Pakola, Santeri; Basnet, Saru; Grönberg-Vähä-Koskela, Susanna; Arias, Victor; Gladwyn-Ng, Ivan; Aro, Katri; Bäck, Leif; Räsänen, Jari; Ilonen, Ilkka; Borenius, Kristian; Räsänen, Mikko; Hemminki, Otto; Rannikko, Antti; Kanerva, Anna; Tapper, Johanna; Hemminki, Akseli (2022)
    Cytokines have proven to be effective for cancer therapy, however whilst low-dose monotherapy with cytokines provides limited therapeutic benefit, high-dose treatment can lead to a number of adverse events. Interleukin 7 has shown promising results in clinical trials, but anti-cancer effect was limited, in part due to a low concentration of the cytokine within the tumor. We hypothesized that arming an oncolytic adenovirus with Interleukin 7, enabling high expression localized to the tumor microenvironment, would overcome systemic delivery issues and improve therapeutic efficacy. We evaluated the effects of Ad5/3-E2F-d24-hIL7 (TILT-517) on tumor growth, immune cell activation and cytokine profiles in the tumor microenvironment using three clinically relevant animal models and ex vivo tumor cultures. Our data showed that local treatment of tumor bearing animals with Ad5/3- E2F-d24-hIL7 significantly decreased cancer growth and increased frequency of tumor-infiltrating cells. Ad5/3-E2F-d24-hIL7 promoted notable upregulation of pro-inflammatory cytokines, and concomitant activation and migration of CD4+ and CD8 + T cells. Interleukin 7 expression within the tumor was positively correlated with increased number of cytotoxic CD4+ cells and IFNg-producing CD4+ and CD8+ cells. These findings offer an approach to overcome the current limitations of conventional IL7 therapy and could therefore be translated to the clinic.
  • Quixabeira, Dafne C. A.; Cervera-Carrascon, Victor; Santos, Joao M.; Clubb, James H. A.; Kudling, Tatiana V.; Basnet, Saru; Heiniö, Camilla; Grönberg-Vähä-Koskela, Susanna; Anttila, Marjukka; Havunen, Riikka; Kanerva, Anna; Hemminki, Akseli (2022)
    Intratumoral immunotherapies are entering clinical use but concerns remain regarding their effects on non-injected tumors. Here, we studied the impact of local treatment with an adenovirus coding for TNFa and IL-2 on systemic antitumor response in animals receiving aPD-1 (anti-programmed cell death protein 1) therapy. Using bilateral murine melanoma models, we tested systemic tumor response to combined therapy with anti-PD-1 and an adenovirus coding for TNFa and IL-2 ("virus"). Virus was given intratumorally (to one of the two tumors only) and aPD-1 monoclonal antibody systemically. We evaluated both tumors' response to treatment, overall survival, metastasis development, and immunological mechanisms involved with response. Consistent tumor control was observed in both injected and non-injected tumors, including complete response in all treated animals receiving aPD-1+ virus therapy. Mechanistically, virus injections enabled potent effector lymphocyte response locally, with systemic effects in non-injected tumors facilitated by aPD-1 treatment. Moreover, adenovirus therapy demonstrated immunological memory formation. Virus therapy was effective in preventing metastasis development. Local treatment with TNFa and IL-2 coding adenovirus enhanced systemic response to aPD-1 therapy, by re-shaping the microenvironment of both injected and non-injected tumors. Therefore, our pre-clinical data support the rationale for a trial utilizing a combination of aPD-1 plus virus for the treatment of human cancer.
  • Sahi, Helka; Their, Jenny; Gissler, Mika; Koljonen, Virve (2020)
    Merkel cell carcinoma (MCC) is a rare cutaneous carcinoma that has gained enormous interest since the discovery of Merkel cell polyoma virus, which is a causative oncogenic agent in the majority of MCC tumours. Increased research has focused on effective treatment options with immuno-oncology. In this study, we reviewed the real-world data on different treatments given to MCC patients in Finland in 1986-2016. We used the Finnish Cancer Registry database to find MCC patients and the Hospital Discharge Register and the Cause-of-Death Register to obtain treatment data. We identified 376 MCC patients and 33 different treatment entities and/or combinations of treatment. An increase was noted in the incidence of MCC since 2005. Therefore, the cohort was divided into two groups: the "early" group with time of diagnosis between years 1986 and 2004 and the "late" group with time of diagnosis between 2005 and 2016. The multitude of different treatment combinations is a relatively new phenomenon; before the year 2005, only 11 treatments or treatment combinations were used for MCC patients. Our data show that combining radiation therapy with simple excision provided a survival advantage, which was, however, lost after adjustment for stage or age. Our registry study serves as a baseline treatment efficacy comparison as we move into the age of immunotherapy in MCC. Standardizing the treatment of MCC patients in Finland requires more work on awareness and multidisciplinary co-operation.
  • Gao, Yan; Tong, Haibei; Li, Jialiang; Li, Jiachen; Huang, Di; Shi, Jisen; Xia, Bing (2021)
    Nanomedicines have been designed and developed to deliver anticancer drugs or exert anticancer therapy more selectively to tumor sites. Recent investigations have gone beyond delivering drugs to tumor tissues or cells, but to intracellular compartments for amplifying therapy efficacy. Mitochondria are attractive targets for cancer treatment due to their important functions for cells and close relationships to tumor occurrence and metastasis. Accordingly, multifunctional nanoplatforms have been constructed for cancer therapy with the modification of a variety of mitochondriotropic ligands, to trigger the mitochondria-mediated apoptosis of tumor cells. On this basis, various cancer therapeutic modalities based on mitochondria-targeted nanomedicines are developed by strategies of damaging mitochondria DNA (mtDNA), increasing reactive oxygen species (ROS), disturbing respiratory chain and redox balance. Herein, in this review, we highlight mitochondria-targeted cancer therapies enabled by nanoplatforms including chemotherapy, photothermal therapy (PTT), photodynamic therapy (PDT), chemodynamic therapy (CDT), sonodynamic therapy (SDT), radiodynamic therapy (RDT) and combined immunotherapy, and discussed the ongoing challenges.